Endometrial hyperplasia and progesterone resistance: a complex relationship

Cover Page

Cite item

Abstract

The endometrium is one of the most dynamic tissues that constantly undergoes changes during the menstrual cycle in women of the reproductive period. All these processes take place mainly under the influence of steroid hormones that are produced in the woman’s body. However, it is important to remember that throughout life the endometrial tissue undergoes changes under the influence of various factors that lead to imbalances in hormonal regulation. All these changes can lead to the development of endometrial hyperplasia, which has a high risk of both recurrence and malignization. Over the past few decades, the incidence of endometrial cancer has increased in many countries. This trend is thought to be related to the increasing prevalence of obesity, as well as to changing female reproductive patterns. Although there are currently no well-established screening programmers for endometrial cancer, endometrial hyperplasia is a recognized precursor, and its detection provides an opportunity for prevention. Studying the pathogenesis and risk factors will give a great advantage in the future to prevent possible complications. At this point, the activity and inhibition of the different hormone isoforms can lead to different hyperplastic processes. The management of patients depends on many factors: age, species, reproductive potential and other factors. Therefore, a comprehensive approach to treatment is always necessary. In recent years, interest in the study of endometrial hyperplasia has increased dramatically due to the increase in endometrial cancer. Therefore, the issue of early diagnosis and prevention is most urgent in modern gynecology and requires further study. This review reflects the current understanding of the disruption of progesterone signaling mechanisms in endometrial hyperplasia according to domestic and foreign literature.

Full Text

Introduction

The endometrium is the inner layer of the uterus, which undergoes a constant cyclical change in women of reproductive age, allowing us to speak of it as one of the most dynamic tissues [1]. Processes such as desquamation, regeneration, proliferation and differentiation occur mainly under the influence of steroid hormones with ovarian genesis. Steroid hormones and their signaling mechanisms are strictly regulated to maintain a normal menstrual cycle. Estrogen in the female body promotes proliferation, but an increased concentration of progesterone inhibits the action of estrogen, causing decidualization [2]. During the reproductive period, the endometrium is exposed to various factors that lead to hormonal insensitivity, hypo/hyperestrogenism, progesterone resistance, i. e. hormonal imbalance. In turn, changes in gene expression and epigenetic markers are more likely to disrupt endometrial tissue regulation, creating a hormone insensitive environment [3–6]. Reduced cell response to progesterone and/or impaired progesterone receptor (PR) activation leads to the development of gynecological diseases, including endometrial hyperplasia (EH) [7–10].

In the age of molecular medicine, there is an urgent need to elucidate the mechanisms leading to the occurrence or progression of gynecological diseases due to impaired signaling transmissions and cellular response to progesterone [11–13]. At this point, modern medicine should focus on identifying the causes of hormonal imbalance, such as gene mutation, and the improper regulation of steroid hormone signaling, which will then lead to the selection of the right management tactics for patients.

One of the main links in the regulation of reproductive functions is progesterone, which has points of application in various organs: uterus, ovaries, mammary glands, brain. Progesterone actions are mediated by progesterone receptors (PR), which consist mainly of two nuclear isoforms (PRA and PRB) with different expression patterns and functional profiles [14].

Progesterone resistance in the endometrium

Progesterone resistance in the endometrium is a pathological condition that leads to dysregulation of epithelial and stromal gene expression in the endometrium [15–18]. These abnormal pathophysiological changes have a cumulative effect, which will subsequently lead to the development of endometrial- related diseases, including endometrial hyperplasia (EH) [15–19]. According to the literature, when studying the gene expression of pathological processes that have a hyperplastic nature, it was found that changes are observed in the early and middle period of the secretory phase. These processes adversely affect the endometrium and are associated with a loss of normal function, leading to further disease progression [20, 21]. It should always be remembered that any proliferative processes may soon lead to malignisation. Epigenetic changes, including hypermethylation, reduce PR expression and lead to progesterone resistance [17, 22]. As previously mentioned, PR isoforms have different functional profiles. Thus, PR-B activates the target gene sites for progesterone, the ‘activating’ isoform, while PR-A acts as an inhibitor of this hormone receptor [14]. The effect of PR isoforms on the development of progesterone resistance was first shown in 2000 by western colleagues [23]. It is worth noting that progesterone- regulated genes, which play an important role in estrogen metabolism (conversion of biologically active estradiol to less potent estrone), also contribute to proliferative endometrial diseases [9].

Thus, any alterations such as gene expression, epigenetic mutations and/or gene mutations are highly likely to affect progesterone signaling in the endometrium.

Endometrial hyperplasia

Endometrial hyperplasia (EH) is a proliferation under the influence of hormonal imbalance that results in increased volume and altered endometrial tissue architectonics, with a change in the endometrial gland- stromal ratio of more than 1:1 [24–26].

In 2014, the World Health Organization (WHO), taking into account the clinical presentation and management of patients, proposed a binary classification of HE with and without atypia [27].

The rate of transformation to cancer varies and is less than 1–3 % for hyperplasia without atypia, and up to 25–29 % for atypical hyperplasia [28, 29]. However, it should also be known that endometrial hyperplasia without atypia has a 7 % risk of atypical endometrial hyperplasia and a 15 % risk of endometrial cancer [30].

Endometrial hyperplastic processes are precursors to malignancy [31, 32]. Adenocarcinoma is the most common endometrial carcinoma, accounting for more than 80 % of all endometrial carcinomas [33–35]. It is well known from the literature that endometrial cancer is correlated with genetic changes in PTEN, KRAS, CTNNB1, ARID1A and PIK3CA. About 65 % of adenocarcinoma development is associated with a PTEN mutation [36]. However, it is worth noting that PTEN mutations are also observed in the development of endometrial hyperplasia [37]. Some authors [38, 39] believe that a PTEN mutation is sufficient to develop uterine corpus cancer, while others [40] suggest that malignancies are cumulative and require different triggers, combinations of mutations that complement each other, such as PTEN KRAS, CTNNB1, ARID1A and PIK3CA, for a more aggressive manifestation. An interesting observation seen by Western colleagues is that PTEN mutations when exposed to oestrogen lead to an increased incidence of endometrial carcinomas [41].

Management tactics  for endometrial hyperplasia

Therapy for endometrial hyperplasia in women aims at stopping bleeding, restoring menstrual function in the reproductive period or achieving endometrial atrophy and subatrophy in the perimenopausal age, and preventing relapse of the hyperplastic process [24].

The management of the patient depends on various factors: age, type of GE, clinical situation, reproductive plans. In recent years, the use of progestins in endometrial hyperplasia and their efficacy in treatment have been studied extensively. According to the literature, the response of progestin therapy is variable, and is associated with heterogeneity of mutations. [1, 32]. It is crucial to understand the pathogenesis of endometrial hyperplasia in order to obtain a favorable outcome to conservative treatment [32].

Conclusion

Endometrial hyperplasia has a different etiology, pathogenesis and is multifactorial in nature. However, the influence of impaired regulation of steroid hormone signaling in the study of pathogenesis cannot be denied. This may be due to imbalances in hormone production, progesterone resistance, altered hormone- dependent gene expression, and common somatic gene mutations. The dynamic changes in the endometrium during the reproductive period represent a complex mechanism that is subject to various influences throughout life. Numerous multicenter studies on the etiology and pathogenesis have contributed to the development of a management algorithm.

×

About the authors

Mekan R. Orazov

Russian People’s Friendship University

Email: 211irina2111@rambler.ru
ORCID iD: 0000-0002-1767-5536
Moscow, Russian Federation

Ludmila M. Mikhaleva

Avtsyn Research Institute of Human Morphology of Federal state budgetary scientific institution «Petrovsky National Research Centre of Surgery»

Email: 211irina2111@rambler.ru
ORCID iD: 0000-0003-2052-914X
Moscow, Russian Federation

Irina A. Mullina

Russian People’s Friendship University

Author for correspondence.
Email: 211irina2111@rambler.ru
ORCID iD: 0000-0002-5773-6399
Moscow, Russian Federation

References

  1. MacLean JA 2nd, Hayashi K. Progesterone Actions and Resistance in Gynecological Disorders. Cells. 2022;11(4):647. doi: 10.3390/cells11040647
  2. Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr Rev. 2014;35(6):851-905. doi: 10.1210/er.2014-1045
  3. Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR, Lessey BA, Giudice LC. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 2007;148(8):3814-26. doi: 10.1210/en.2006-1692
  4. Kao LC, Germeyer A, Tulac S, Lobo S, Yang JP, Taylor RN, Osteen K, Lessey BA, Giudice LC. Expression profiling of endometrium from women with endometriosis reveals candidate genes for disease-based implantation failure and infertility. Endocrinology. 2003;144(7):2870-81. doi: 10.1210/en.2003-0043
  5. Houshdaran S, Nezhat CR, Vo KC, Zelenko Z, Irwin JC, Giudice LC. Aberrant Endometrial DNA Methylome and Associated Gene Expression in Women with Endometriosis. Biol Reprod. 2016;95(5):93. doi: 10.1095/biolreprod.116.140434
  6. Houshdaran S, Oke AB, Fung JC, Vo KC, Nezhat C, Giudice LC. Steroid hormones regulate genome-wide epigenetic programming and gene transcription in human endometrial cells with marked aberrancies in endometriosis. PLoS Genet. 2020;16(6): e1008601. doi: 10.1371/journal.pgen.1008601
  7. Al- Sabbagh M, Lam EW, Brosens JJ. Mechanisms of endometrial progesterone resistance. Mol Cell Endocrinol. 2012;358(2):208-15. doi: 10.1016/j.mce.2011.10.035.
  8. McKinnon B, Mueller M, Montgomery G. Progesterone Resistance in Endometriosis: an Acquired Property? Trends Endocrinol Metab. 2018;29(8):535-548. doi: 10.1016/j.tem.2018.05.006
  9. Patel BG, Rudnicki M, Yu J, Shu Y, Taylor RN. Progesterone resistance in endometriosis: origins, consequences and interventions. Acta Obstet Gynecol Scand. 2017;96(6):623-632. doi: 10.1111/aogs.13156
  10. Li X, Feng Y, Lin JF, Billig H, Shao R. Endometrial progesterone resistance and PCOS. J Biomed Sci. 2014;21(1):2. doi: 10.1186/1423-0127-21-2
  11. DeMayo FJ, Zhao B, Takamoto N, Tsai SY. Mechanisms of action of estrogen and progesterone. Ann N Y Acad Sci. 2002;955:48-59; discussion 86-8, 396-406. doi: 10.1111/j.1749-6632.2002.tb02765.x
  12. Kim JJ, Kurita T, Bulun SE. Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer. Endocr Rev. 2013;34(1):130-62. doi: 10.1210/er.2012-1043
  13. Marquardt RM, Kim TH, Shin JH, Jeong JW. Progesterone and Estrogen Signaling in the Endometrium: What Goes Wrong in Endometriosis? Int J Mol Sci. 2019;20(15):3822. doi: 10.3390/ijms20153822
  14. Patel B, Elguero S, Thakore S, Dahoud W, Bedaiwy M, Mesiano S. Role of nuclear progesterone receptor isoforms in uterine pathophysiology. Hum Reprod Update. 2015;21(2):155-73. doi: 10.1093/humupd/dmu056
  15. Al- Sabbagh M, Lam EW, Brosens JJ. Mechanisms of endometrial progesterone resistance. Mol Cell Endocrinol. 2012;358(2):208-15. doi: 10.1016/j.mce.2011.10.035
  16. McKinnon B, Mueller M, Montgomery G. Progesterone Resistance in Endometriosis: an Acquired Property? Trends Endocrinol Metab. 2018;29(8):535-548. doi: 10.1016/j.tem.2018.05.006
  17. Patel BG, Rudnicki M, Yu J, Shu Y, Taylor RN. Progesterone resistance in endometriosis: origins, consequences and interventions. Acta Obstet Gynecol Scand. 2017;96(6):623-632. doi: 10.1111/aogs.13156
  18. Li X, Feng Y, Lin JF, Billig H, Shao R. Endometrial progesterone resistance and PCOS. J Biomed Sci. 2014;21(1):2. doi: 10.1186/1423-0127-21-2
  19. Moustafa S, Young SL. Diagnostic and therapeutic options in recurrent implantation failure. F1000Res. 2020;9: F1000 Faculty Rev-208. doi: 10.12688/f1000research.22403.1
  20. Savaris RF, Groll JM, Young SL, DeMayo FJ, Jeong JW, Hamilton AE, Giudice LC, Lessey BA. Progesterone resistance in PCOS endometrium: a microarray analysis in clomiphene citrate-treated and artificial menstrual cycles. J Clin Endocrinol Metab. 2011;96(6):1737-46. doi: 10.1210/jc.2010-2600
  21. Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR, Lessey BA, Giudice LC. Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 2007;148(8):3814-26. doi: 10.1210/en.2006-1692
  22. Guo SW. Epigenetics of endometriosis. Mol Hum Reprod. 2009;15(10):587-607. doi: 10.1093/molehr/gap064
  23. Attia GR, Zeitoun K, Edwards D, Johns A, Carr BR, Bulun SE. Progesterone receptor isoform A but not B is expressed in endometriosis. J Clin Endocrinol Metab. 2000;85(8):2897-902. doi: 10.1210/jcem.85.8.6739
  24. Orazov MR, Hamoshina MB, Mullina IA, Artemenko JS. Endometrial hyperplasia - from pathogenesis to effective therapy. Obstetrics and gynaecology: news, opinions, training. 2021;9(3):21-28. (In Russian).
  25. Kim JJ, Chapman- Davis E. Role of progesterone in endometrial cancer. Semin Reprod Med. 2010;28(1):81-90. doi: 10.1055/s-0029-1242998
  26. Singh G, Puckett Y. Endometrial Hyperplasia. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing. 2022. 461 p.
  27. Emons G, Beckmann MW, Schmidt D, Mallmann P; Uterus commission of the Gynecological Oncology Working Group (AGO). New WHO Classification of Endometrial Hyperplasias. Geburtshilfe Frauenheilkd. 2015;75(2):135-136. doi: 10.1055/s-0034-1396256
  28. Ørbo A, Arnes M, Vereide AB, Straume B. Relapse risk of endometrial hyperplasia after treatment with the levonorgestrel-impregnated intrauterine system or oral progestogens. BJOG. 2016;123(9):1512-9. doi: 10.1111/1471-0528.13763
  29. Erdem B, Aşıcıoğlu O, Seyhan NA, Peker N, Ülker V, Akbayır Ö. Can concurrent high-risk endometrial carcinoma occur with atypical endometrial hyperplasia? Int J Surg. 2018;53:350-353. doi: 10.1016/j.ijsu.2018.04.019
  30. Iversen ML, Dueholm M. Complex non atypical hyperplasia and the subsequent risk of carcinoma, atypia and hysterectomy during the following 9-14 years. Eur J Obstet Gynecol Reprod Biol. 2018;222:171-175. doi: 10.1016/j.ejogrb.2018.01.026
  31. Li L, Yue P, Song Q, Yen TT, Asaka S, Wang TL, Beavis AL, Fader AN, Jiao Y, Yuan G, Shih IM, Song Y. Genome-wide mutation analysis in precancerous lesions of endometrial carcinoma. J Pathol. 2021;253(1):119-128. doi: 10.1002/path.5566
  32. Pandey J, Yonder S. Premalignant Lesions Of The Endometrium. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing. 2022.
  33. Huvila J, Pors J, Thompson EF, Gilks CB. Endometrial carcinoma: molecular subtypes, precursors and the role of pathology in early diagnosis. J Pathol. 2021;253(4):355-365. doi: 10.1002/path.5608
  34. Carugno J, Marbin SJ, LaganÀ AS, Vitale SG, Alonso L, DI Spiezio Sardo A, Haimovich S. New development on hysteroscopy for endometrial cancer diagnosis: state of the art. Minerva Med. 2021;112(1):12-19. doi: 10.23736/S0026-4806.20.07123-2
  35. Yen TT, Wang TL, Fader AN, Shih IM, Gaillard S. Molecular Classification and Emerging Targeted Therapy in Endometrial Cancer. Int J Gynecol Pathol. 2020;39(1):26-35. doi: 10.1097/PGP.0000000000000585
  36. Cancer Genome Atlas Research Network, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, Robertson AG, Pashtan I, Shen R, Benz CC, Yau C, Laird PW, Ding L, Zhang W, Mills GB, Kucherlapati R, Mardis ER, Levine DA. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67-73. doi: 10.1038/nature12113
  37. Ayhan A, Mao TL, Suryo Rahmanto Y, Zeppernick F, Ogawa H, Wu RC, Wang TL, Shih Ie M. Increased proliferation in atypical hyperplasia/endometrioid intraepithelial neoplasia of the endometrium with concurrent inactivation of ARID1A and PTEN tumour suppressors. J Pathol Clin Res. 2015;1(3):186-93. doi: 10.1002/cjp2.22
  38. Joshi A, Miller C Jr, Baker SJ, Ellenson LH. Activated mutant p110α causes endometrial carcinoma in the setting of biallelic Pten deletion. Am J Pathol. 2015;185(4):1104-13. doi: 10.1016/j.ajpath.2014.12.019
  39. Kim TH, Wang J, Lee KY, Franco HL, Broaddus RR, Lydon JP, Jeong JW, Demayo FJ. The Synergistic Effect of Conditional Pten Loss and Oncogenic K-ras Mutation on Endometrial Cancer Development Occurs via Decreased Progesterone Receptor Action. J Oncol. 2010;2010:139087. doi: 10.1155/2010/139087
  40. Suryo Rahmanto Y, Shen W, Shi X, Chen X, Yu Y, Yu ZC, Miyamoto T, Lee MH, Singh V, Asaka R, Shimberg G, Vitolo MI, Martin SS, Wirtz D, Drapkin R, Xuan J, Wang TL, Shih IM. Inactivation of Arid1a in the endometrium is associated with endometrioid tumorigenesis through transcriptional reprogramming. Nat Commun. 2020;11(1):2717. doi: 10.1038/s41467-020-16416-0
  41. Joshi A, Wang H, Jiang G, Douglas W, Chan JS, Korach KS, Ellenson LH. Endometrial tumorigenesis in Pten(+/-) mice is independent of coexistence of estrogen and estrogen receptor α. Am J Pathol. 2012;180(6):2536-47. doi: 10.1016/j.ajpath.2012.03.006

Copyright (c) 2023 Orazov M.R., Mikhaleva L.M., Mullina I.A.

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies